F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

REVIEW

Recent advances in understanding multiple myeloma [version 1; referees: 4 approved] Binod Dhakal1, Saulius Girnius2, Parameswaran Hari1 1Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA 2Division of Hematology/Oncology, University of Cincinnati, Cincinnati, OH, USA

v1

First published: 23 Aug 2016, 5(F1000 Faculty Rev):2053 (doi: 10.12688/f1000research.8777.1)

Open Peer Review

Latest published: 23 Aug 2016, 5(F1000 Faculty Rev):2053 (doi: 10.12688/f1000research.8777.1)

Referee Status:

Abstract There have been major recent advancements in the understanding and management of multiple myeloma. Diagnostic criteria have been revised and former ultra-high-risk smoldering multiple myeloma is now considered multiple myeloma in need of treatment. Understanding clonal progression, evolution, and tides not only has helped elucidate the disease behavior but might help expand therapeutic choices in order to select appropriate treatment for patients. Unprecedented response rates with modern triplet induction therapies containing proteasome inhibitor and immunomodulators have made this approach standard for initial treatment. The US Food and Drug Administration approved four new drugs (two targeted antibodies and two oral agents) in 2015 in relapsed/refractory multiple myeloma and these drugs along with the other already-available drugs have now increased the choices of regimens. Even drugs without single-agent activity, such as panobinostat and elotuzumab, have an important role, especially in the proteasome inhibitor refractory setting. Recent studies done in the context of novel agent induction suggest that high-dose therapy followed by autologous transplant continues to improve response rates and progression-free survival, thus underscoring their role in transplant-eligible patients. Evolving paradigms in the treatment of multiple myeloma include newer promising immune approaches, such as adoptive cellular therapies, vaccines, or antibody-based immune manipulations. Though multiple myeloma is still considered incurable, it is clear that with the improved understanding of disease biology and clonal architecture of relapse combined with the availability of multi-targeted approaches, we are ever closer to a lasting cure or transformation into indolent and long-lasting disease courses or both.

Invited Referees

1

2

3

4

version 1 published 23 Aug 2016

F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty. In order to make these reviews as comprehensive and accessible as possible, peer review takes place before publication; the referees are listed below, but their reports are not formally published. 1 Monika Engelhardt, University of Freiburg Medical Center Germany 2 Stefan Knop, Wuerzburg University Medical Center Germany 3 Qaiser Bashir, The University of Texas M.D. Anderson Cancer Center USA 4 David Avigan, Beth Israel Deaconess Medical Center & Harvard Medical School USA

Discuss this article Comments (0)

F1000Research Page 1 of 10

F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

Corresponding author: Parameswaran Hari ([email protected]) How to cite this article: Dhakal B, Girnius S and Hari P. Recent advances in understanding multiple myeloma [version 1; referees: 4 approved] F1000Research 2016, 5(F1000 Faculty Rev):2053 (doi: 10.12688/f1000research.8777.1) Copyright: © 2016 Dhakal B et al. This is an open access article distributed under the terms of the Creative Commons Attribution Licence, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Grant information: The author(s) declared that no grants were involved in supporting this work. Competing interests: The authors declare that they have no competing interests. First published: 23 Aug 2016, 5(F1000 Faculty Rev):2053 (doi: 10.12688/f1000research.8777.1)

F1000Research Page 2 of 10

F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

Introduction Multiple myeloma (MM) is a clonal expansion of malignant plasma cells resulting in end organ damage, including lytic bone lesions, anemia, renal failure, or hypercalcemia. Both the understanding of MM and its management have evolved rapidly over recent years. Concomitant with the increasing incidence of MM to almost 30,000 patients annually in the US, mortality has trended down in the last decade, resulting in an increased prevalence of MM1. In 2015, four new drugs—the first two monoclonal antibodies and two oral agents—were approved by the US Food and Drug Administration (FDA). Importantly, these drugs have improved the outcomes in relapsed and heavily pre-treated patients.

(c) more than one focal lesion on magnetic resonance imaging. Each of these biomarkers was associated with an 80% risk of progression to symptomatic MM in 2 years in independent studies4,7,8. Staging of MM was recently updated to reflect the advances in treatment of MM as well as improved detection of high-risk disease. Revised-International Staging System (R-ISS) stage I disease includes ISS stage I with a normal lactate dehydrogenase (LDH) level and absence of high-risk cytogenetics (del(17p), t(4;14), or t(14;16)). R-ISS III includes ISS stage III with either high-risk cytogenetics or elevated LDH. R-ISS II includes all others. The 5-year OS rates were 82% in R-ISS I, 62% in R-ISS II, and 40% in R-ISS III12.

High-risk smoldering multiple myeloma Smoldering MM (SMM) was traditionally defined on the basis of laboratory criteria—monoclonal (M) protein level of more than 3 g/dl or a clonal marrow plasmacytosis of more than 10%—and the absence of end organ damage. The ability to identify persons with SMM who are likely to progress rapidly to symptomatic MM has led to the revision of the criteria that define treatment initiation. Ultra-high-risk SMM patients with an 80% risk of progression to MM in 2 years are now considered in the updated criteria to have MM requiring treatment2. Currently, SMM is defined by the presence of serum M protein of at least 3 g/dl or 10 to 60% of clonal bone marrow plasma cells (or both) with no evidence of target organ damage, myeloma-defining events (MDEs), or amyloidosis2. Patients with high-risk SMM have a 25% risk of progression to MM in 2 years, whereas those with low risk have a 5% risk per year3. Multiple prognostic markers have been identified as surrogates for high-risk SMM4–9. Some of these include SMM with serum M protein of at least 3 g/dl, IgA isotype, involved/uninvolved free light chain ratio of at least 8 (but less than 100), bone marrow clonal plasma cells of 50 to 60%, t(4;14), del(17p), or 1q gain and Bence-Jones proteinuria. Management of high-risk SMM remains controversial after treatment with lenalidomide was shown to improve overall survival (OS) compared with a strategy of observation until target organ damage10. It is possible that SMM represents a curable stage of disease wherein early intervention approaches with limited toxicity may be curative and delay or avoid end organ damage provided that treatment does not lead to the outgrowth of more resistant clones. When compared with symptomatic MM, high-risk SMM can achieve a deeper response, as measured by minimal residual disease (MRD), with 11 out of 12 patients developing MRD negativity11. A longer follow-up is necessary to determine the duration of response, comparing it with the financial and physical burden of therapy. Thus, these treatments should be performed in clinical trials exploring the timing, the benefits, and the type of therapy in high-risk SMM.

New criteria for multiple myeloma The revised International Myeloma Working Group criteria for the diagnosis of MM requires, in addition to the presence of either 10% or more clonal plasma cells or a biopsy-proven plasmacytoma, one or more MDEs. MDE includes CRAB features (increased calcium level, renal dysfunction, anemia, and destructive bone lesions) and three specific biomarkers: (a) clonal plasma cells of at least 60%, (b) involved/uninvolved light chain ratio of at least 100, and

Clonal dominance The initial event leading to malignant plasma cells occurs in the germinal center during isotype class switching and somatic hypermutation. These mutations are present in monoclonal gammopathies of unknown significance but are not sufficient for evolution to MM13. The progression to MM involves additional oncogenic events, including activation of oncogenes K-Ras, N-Ras, FAM46C, MYC, and BRAF and loss of function of p53, epigenetic changes with histone methylation/deacetylation, and genomic instability14,15. No one common driver mutation has been identified in MM, although recurrent variants have been identified. Deregulation of the RAS/MAPK, NF-κB pathway, and apoptotic response are involved, but only the inability for effective apoptotic response to DNA damage has prognostic value15. A second study confirmed that the presence of the aforementioned oncogenes did not influence OS or progression-free survival (PFS)16. In MM, at least four distinct evolutionary patterns can be discerned between initial treatment phase and subsequent relapses. The simplest situation is no change in the clonal and sub-clonal composition between various time points in the natural course of disease, which represents re-emergence of the original clonal plasma cell population at relapses. About one-third of patients in this group have stable genomes and are typically associated with hyperploidy and favorable outcomes. The other three scenarios include “either differential clonal response or clonal tides” where in subclones are identified at different time points, but their relative proportions change over time or linear clonal evolution where a new (previously undetectable) sub-clone emerges later or a branching evolution pattern corresponding to the emergence of new clones with the disappearance of old ones16,17. In these three groups, selective therapeutic pressures can suppress responsive clones, allowing resistant clones to become dominant. Persons with high-risk disease are more likely to exhibit linear evolution or clonal tides. Clones with inherent genomic instability, detected at low levels at diagnosis, may undergo further genomic shifts when exposed to DNA-damaging agents, emerging after multiple relapses as the fatal clone. Understanding the clonal evolution pattern will become increasingly relevant as therapeutic choices expand in order to select appropriate treatment for patients. Re-emergence of a previously sensitive sub-clone would warrant the use of the same combination therapy at relapse. Alternatively, the emergence under treatmentinduced selection pressure of a newly dominant linearly evolved or pre-existing resistant sub-clone should lead to alternate drugs.

Page 3 of 10

F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

Newer proteasome inhibitors Two new proteasome inhibitors (PIs) have been approved in recent years: carfilzomib (K) and ixazomib (I). Carfilzomib is an intravenous, irreversible PI with impressive outcomes in both newly diagnosed MM and relapsed MM. In patients with early relapse (one to three prior regimens), when compared with lenalidomide and dexamethasone (RD), carfilzomib in combination with lenalidomide and dexamethasone (KRD) improved PFS (hazard ratio (HR) 0.69, P = 0.0001) and OS (HR 0.79, P = 0.04)18. In newly diagnosed patients, the KRD triplet had a response rate of 98%, a complete response rate of 56%, and MRD negativity rate by next-generation sequencing of 58%, and no discontinuation due to adverse events11. Benefits of carfilzomib include rapid time to response, excellent tolerability (15% discontinued due to adverse events), efficacy in high-risk disease, and the low rate of carfilzomib-induced peripheral neuropathy. Challenges with carfilzomib include twice-weekly intravenous administration, the risk for tumor lysis syndrome, and the possibility for cardiac events and dyspnea. One retrospective study identified that more than 20% of patients developed systolic heart failure or arrhythmias. Previous irradiation to the thoracic spine and concomitant administration of doxorubicin increased the risk for cardiac dysfunction19. To overcome these challenges, recent trials have demonstrated tolerability and response with escalated doses20, whereas others are investigating efficacy of once-weekly dosing. Ixazomib is an orally bioavailable, reversible PI with higher tissue distribution and a shorter dissociation half-life than bortezomib21. It has modest single-agent activity and response to bortezomibexposed persons when combined with dexamethasone22. The combination of ixazomib with lenalidomide and dexamethasone (IRD) compared with RD shortened the time to response, improved the overall response rate, and increased median PFS (20.6 versus 14.7 months, HR 0.74, P = 0.012)23. Early signal also suggests that it may overcome the negative impact of cytogenetic alterations. Major toxicities include gastrointestinal (GI) toxicity, peripheral neuropathy, and cyclical thrombocytopenia. The tissue distribution and dissociation half-life have a theoretical benefit with extramedullary disease, although this has not yet been demonstrated in a clinical trial. Ixazomib can be used as the proteasome backbone for combination oral therapies, reducing the need for frequent infusion visits. IRD is being investigated in newly diagnosed, transplant-ineligible persons with MM in a multi-center clinical trial, but results are not yet available. The availability of an oral PI also increases the feasibility of oral PI-based maintenance/consolidation regimens after transplant. Although it has been approved by the FDA, European regulators are waiting for additional positive data.

Immunomodulatory drug Immunomodulatory drug (IMID) can bind cereblon on the E3 ubiquitin ligase complex, enhancing the ubiquitination and degradation of two zinc finger transcription factors24,25. Pomalidomide is an oral third-generation IMID approved for relapsed refractory MM after at least two prior regimens. In lenalidomide-refractory patients, including 75% who were dual refractory, the response rate for pomalidomide and weekly dexamethasone was 30% with a 4-month PFS26. In high-risk cytogenetics, patients with del(17p) had higher response rates, duration of response, and time to progression

than those with t(4;14)27. Pomalidomide is more effective earlier in the disease course28, although approximately 40% of bortezomiband lenalidomide-refractory patients can have a prolonged PFS29. Toxicities, including cytopenias, rash, and peripheral neuropathy, are unpredictable30. Unlike prior IMIDs, pomalidomide causes less endothelial stress and may be associated with lower rates of venous thromboembolic events, although patients should be on prophylactic anti-platelet therapy or anticoagulation31. Its limited toxicity allows for combination with PIs, cyclophosphamide, and monoclonal antibodies, improving its efficacy.

Histone deacetylase inhibitor Panobinostat, a pan-histone deacetylase inhibitor (HDAC), targets HDAC enzymes involved in the aggresome protein degradation pathway, working synergistically with PIs. In a heavily pre-treated patient population, including 73% who were bortezomib-refractory, the response rate of panobinostat, bortezomib, and dexamethasone was 35% with a 5.4-month PFS32. In bortezomib-refractory patients, the clinical benefit rate was better than 50% with a 4.2-month PFS. As high rates of GI distress and thrombocytopenia are seen with this agent, tolerability is a major impediment to its use and requires intense clinic/nursing support for continuous use. Dose interruptions occur in more than 50%, and relative dose intensity is only 73%. In a less heavily pre-treated population, response rates and PFS are improved, but the difficulty in maintaining dose intensity persists33. Nonetheless, panobinostat has a novel mechanism of action, is an oral agent, and continues to have an important role in the management of MM, especially in the PI-refractory setting.

Monoclonal antibody The FDA has recently approved two monoclonal antibodies in relapsed or refractory MM (RRMM) or both—elotuzumab and daratumumab—which target the signaling lymphocytic activation molecule F7 (SLAMF7) and CD38, respectively34,35. In a phase III trial of patients with RRMM, the addition of elotuzumab to RD significantly improved PFS (19.4 versus 14.9 months, P < 0.001)35. In addition, it improves the PFS of the subsequent therapy, suggesting a lasting effect on natural killer (NK) cells. Single-agent daratumumab produced a response rate of approximately 30% in heavily pre-treated patients and little toxicity other than infusion reactions34. Another CD38 antibody, isatuximab (SAR650984), has shown similar single-agent activity in RRMM but is not commercially available36. Both elotuzumab and daratumumab are being investigated in the front-line setting, and given the unprecedented single-agent activity, it is likely that daratumumab might be the backbone of MM treatment, akin to rituximab in B-cell lymphomas. Emerging data point to impressive responses for anti-CD38 antibodies in combination with IMIDs and PIs.

Triplets at induction When compared with doublets, triplet induction therapy containing a PI and an IMID results in a higher response rate and improved PFS and is preferred whenever possible in fit patients, especially given the synergism between these two drug classes37,38. A recent study has shown that bortezomib-lenalidomide and dexamethasone (VRD) has superior OS compared with RD39. Bortezomib-thalidomide and dexamethasone (VTD) has shown superior response rates than thalidomide and dexamethasone (TD) and bortezomib-dexamethasone

Page 4 of 10

F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

in randomized trials37. Bortezomib-cyclophosphamide and dexamethasone (VCD) was recently shown to have lower response rates than VTD40 (Table 1). Although greater hematologic toxicity is seen in the former and peripheral neuropathy in the latter, the overall rate of grade 3/4 toxicity is similar, and individualization of therapy should dictate treatment decisions41. The second-generation PI carfilzomib in combination with lenalidomide and dexamethasone (KRD) has also shown excellent response rates in phase II trials42, but additional data and regulatory approval are necessary prior to broader use. Conversely, lenalidomide-based triplets failed to show a PFS or OS benefit over lenalidomide-based doublets in an elderly population43. When compared with doublets, triplet therapies are associated with increased toxicity and usually longer PFS but not necessarily OS. The upfront use of the most active agents may limit treatment choices in future lines of therapy, possibly shortening the second and subsequent PFS periods. Given the prolonged survival in patients with myeloma as well as rapidly changing treatment modalities, this debate may be difficult to resolve.

Triplets at first relapse Management of RRMM remains controversial, although the field is moving toward combination triplet therapy at first relapse in fit patients. In the US, an abundance of second-line therapies are available, allowing individualization of therapy to both the patient and the disease. In older persons with RRMM, a geriatric assessment should be performed to determine a frailty score, evaluating the Katz Activities of Daily Living, the Lawton Instrumental Activity of Daily Living, and the Charlson Comorbidity Index44. Frail patients have shorter OS irrespectively of staging, treatment types, or physician-assessed performance status. Even when adjusted for International Staging System and chromosomal abnormalities, frail patients have shorter OS and PFS and higher rates of non-hematologic adverse events and treatment discontinuation and are likely to benefit from dose-adjusted therapy. Other validated frailty indexes evaluate similar factors as well as cytogenetics, renal function, and Karnofsky Performance Status45. The addition of a third drug to an IMID steroid combination improves response rates and PFS, although an OS benefit is not demonstrated consistently18,35,46. The addition of bortezomib

(intravenous) to TD prolongs time to progression (HR 0.59, P = 0.001) in previously transplanted patients and results in a trend toward improved 24-month survival (71% versus 65%, P = 0.093)46. However, this population is not typical for the US, since only 20% were previously treated with bortezomib and 8% with thalidomide. Although VTD may have a limited role in the US, this study does illustrate the benefit of triplet therapy. The addition of carfilzomib to lenalidomide and dexamethasone improves not only response rates and PFS (HR 0.069, P = 0.0001) but also 24-month survival (HR 0.79, P = 0.04)11. The data from other recent trials of triplets in early relapsed disease, though clearly demonstrating PFS benefit, did not show an OS advantage.

Which triplet to use The choice of treatment at relapse is determined by the type, response, and duration of initial therapy as well as disease factors, patient factors, and patient preference. The ideal sequence of therapies is not known, but switching drug classes is likely beneficial, especially in higher-risk disease, given the evidence for clonal tides. For instance, if there is disease progression while on lenalidomide maintenance, a triplet containing lenalidomide would likely provide a suboptimal response. If available, carfilzomib/IMID triplet should be considered in aggressive relapses, given the excellent data with rapid induction of response and high rates of MRD negativity. Those who relapse following a short duration of remission after upfront therapy, including autologous stem cell transplant, or symptomatic relapses with significant clinical symptoms or disease relapse in high-risk cytogenetics are generally considered to be aggressive relapses. Elotuzumab-based treatment could be considered in individuals who are frail or have underlying cytopenias. An ixazomib-based regimen is an option if an oral regimen is desired and, based on early trends, may be considered, especially in del(17p) or extramedullary disease. A pomalidomide-based regimen could also be considered in del(17p). Panobinostat has a limited role in first relapse because of the toxicity profile unless used to restore PI sensitivity in those relapsing after prior PI regimens.

Transplant still better High-dose therapy followed by autologous stem cell transplantation (ASCT) was the standard of care for transplant-eligible patients

Table 1. Triplet induction regimens and autologous stem cell transplantation studies in newly diagnosed multiple myeloma. Number of patients

Overall response rate

PFS

OS

Comments

241 vs. 239

31% vs. 11%

NR

NR

NR

VTD vs. VCD

170 vs. 170

92.3% vs. 83.4%

NR

NR

NR

VRD vs. RD40

242 vs. 232

71% vs. 63%

43 vs. 31 months

NR vs. 63 months

Median OS/PFS

Conventional chemo vs. ASCT48

100 vs. 100

81% vs. 22%

28% vs. 10%

52% vs. 12%

Pre-novel era; 5-year OS/PFS

ASCT vs. MPR57

141 vs. 132

NR

54.7 vs. 37.4 months

78.6% vs. 66.6%

Novel era PFS (median) OS (5-year)

Regimen VTD vs. TD38 42

ASCT, autologous stem cell transplantation; MPR, melphalan/prednisone and revlimid; NR, not reported; OS, overall survival; PFS, progression-free survival; RD, lenalidomide and dexamethasone; TD, thalidomide and dexamethasone; VCD, bortezomib-cyclophosphamide and dexamethasone; VRD, bortezomib-lenalidomide and dexamethasone; VTD, bortezomib-thalidomide and dexamethasone. Page 5 of 10

F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

before the era of novel agents. In pre-novel agent randomized trials, ASCT was uniformly associated with improved response rates and PFS, and about half of the studies showed an OS benefit and superior quality of life47,48. In one of the studies designed to address the question of early versus late ASCT49, patients undergoing ASCT were found to have longer time without symptoms and treatment, and thus the patients with ASCT were found to have improved quality of life. Given the unprecedented response rates with novel agents, the role of ASCT as front-line therapy has been questioned50–52. Novel agents have significantly improved the rate of complete response and MRD, both surrogates for improved survival53,54. The early versus late ASCT was investigated in a number of randomized trials55,56, most recently in an international study conducted in the context of modern therapy with VRD induction, ASCT, and lenalidomide maintenance57. Superior OS benefit was seen in one study56 whereas in the other two studies, early ASCT was found to have better PFS with no transplant-related deaths55,57. Maintenance therapy after ASCT is gaining increased acceptance. Two randomized trials demonstrated a PFS benefit to lenalidomide maintenance, but only the Cancer and Leukemia Group B (CALGB) trial demonstrated an OS benefit58,59. The difference in results could be explained by the use of two cycles of consolidation chemotherapy after ASCT and only one year of maintenance lenalidomide in the IFM (Intergroupe Francophone du Myelome) trial. In addition, the majority of subjects in the CALGB study, unlike the IFM trial, received a lenalidomide-based induction regimen. In addition, an unplanned subset analysis in the CALGB trial failed to show an OS benefit in those who did not receive a lenalidomide-based induction regimen. Likewise, bortezomib maintenance can improve PFS and possibly OS60. Further support for maintenance comes from the observation that maintenance lenalidomide, despite ASCT and consolidation therapy, can further deepen response, when measuring for MRD by flow61. Last, newer data demonstrate the benefit to continuous therapy. A meta-analysis of three trials, one of which included ASCT as part of consolidation, demonstrated improved PFS1, PFS2 (time to second progression of death), and OS with continuous therapy. In fact, a prolonged PFS1 did not reduce PFS262. In conclusion, novel agent-based triplet induction, front-line early ASCT, and maintenance therapy remain the preferred strategy for transplant-eligible patients.

Immunotherapy The best-established immunotherapy in MM is allogeneic transplantation with a potent and often sustained graft-versus-myeloma (GVM) effect63. A series of prospective randomized studies, both in the US and Europe, explored the role of a tandem ASCT and reduced-intensity conditioning allogeneic stem cell transplants from a matched sibling or unrelated donor64,65. The results of these studies have been discordant, especially in the front-line treatment of MM65–68. Nonetheless, younger eligible patients with well-defined high-risk features should be considered for allogeneic transplant, especially within a clinical trial and early in the clinical course (front-line or first relapse). The risks of chronic graft-versus-host disease (cGVHD) and the need for long-term immunosuppression

remain major challenges. A number of studies suggest that immune therapy works in MM and these include demonstration of clinically meaningful GVM effect by donor lymphocyte infusions in patients relapsing after allogeneic transplant69, complete remission after immunosuppression withdrawal62, progression after allogeneic transplant, and the presence of cGVHD correlating with freedom from progression after allogeneic transplant64. A variety of novel post-ASCT or allogeneic transplant immunotherapeutic strategies are now being explored in MM. These include cellular approaches such as myeloma-specific T cells (via T-cell expansion), marrow-infiltrating lymphocytes70, redirected T cells with chimeric antigen receptors71,72, and tumor-based vaccines to induce myeloma-specific immunity in the context of enhanced antigen presentation73. A phase I/II study showed that adoptive transfer of tumor antigen vaccine-primed and co-stimulated T cells leads to augmented and accelerated cellular and humoral immune reconstitution, including antitumor immunity, after ASCT74. In another phase II trial, idiotype-pulsed antigen-presenting cells immunotherapeutic called APC8020 (Mylovenge), which was given after ASCT for MM, appeared to be associated with improved OS75. In a recent report, deep sustained response to anti-CD19 chimeric antigen receptor T cells was reported in a patient with relapsed/ refractory disease76. Several promising antigenic targets have been identified for the development of anti-MM chimeric antigen receptors, such as B-cell maturation antigen, CD138, kappa light chains, and CS-1. Inhibitors of PD-1 and PDL-1 are being studied as a means of breaking down MM immune tolerance71,77. Though the anti-PD1 agent nivolumab was unimpressive78, promising response rates were observed in combination with lenalidomide79. NK cell therapy has generated interest in the treatment of MM and a number of studies are exploring this option. These include modulation of NK activity using anti-KIR Ab IPH2101 (monoclonal antibody against inhibitory KIR on NK cells) to establish MM-specific immunity80 safety and efficacy study of autologous-, allogeneic-, and cord-derived NK cells81,82. A clinical trial involving the haploidentical allo-stem cell transplant followed by planned NK cell infusion attempts to use donor-recipient KIR ligand mismatch and NK cell reactivity to facilitate long-term remission (NCT02100891). Another promising area is the use of tumor vaccine based on the study in which patients receiving a patient-specific dendritic cell/myeloma fusion vaccine demonstrated the expansion of MM-specific T cells as well as upgrading of response in a subgroup of patients73. A randomized phase II trial (BMT CTN 1401) with ASCT followed by lenalidomide maintenance with or without vaccination using the dendritic cell/myeloma fusion vaccine is currently accruing patients. Hematopoietic cell transplantation provides an ideal platform for additional immune-based therapies. The recovery phase from ASCT (or other lymphodepleting therapy) represents a favorable platform for adoptive cellular therapy. The homeostatic lymphocyte proliferation following lymphopenia is a context in which immune checkpoint blockers may also be able to reverse MM-associated T-cell exhaustion71,79. Additionally, lymphopenia resulting from ASCT facilitates elimination of tolerogenic antigen-presenting cells and induces cytokine release that generates a more favorable

Page 6 of 10

F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

environment for adoptive T-cell therapy. Indirect evidence suggests that the immune system can contribute to the clinical benefits of ASCT (for example, those with early lymphoid recovery after ASCT have superior long-term outcomes)83.

number of drug classes as well as new drugs in each class, the combination and sequencing of drugs are largely unresolved.

Conclusions

Competing interests The authors declare that they have no competing interests.

Although MM has always been considered incurable, many investigators now believe that a significant fraction of patients currently undergoing therapy may be cured84. It is clear that with the improved understanding of disease biology and clonal architecture of relapse, combined with the availability of multi-targeted approaches, we are closer to a lasting cure than ever before. Nonetheless, many questions remain unanswered. Will the better understanding of myeloma biology guide the intensity of treatment, with highrisk disease necessitating aggressive therapy? With an increasing

Grant information The author(s) declared that no grants were involved in supporting this work. Acknowledgment We thank Monika Englehardt for the careful critical review of the manuscript.

F1000 recommended

References 1.

Howlader N, Noone AM, Krapcho M, et al.: SEER Cancer Statistics Review (CSR) 1975–2013. National Cancer Institute. Bethesda, MD; 4/2016. Reference Source

2.

Rajkumar SV, Dimopoulos MA, Palumbo A, et al.: International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014; 15(12): e538–48. PubMed Abstract | Publisher Full Text | F1000 Recommendation

myeloma. Nat Rev Cancer. 2012; 12(5): 335–48. PubMed Abstract | Publisher Full Text 14.

Bianchi G, Munshi NC: Pathogenesis beyond the cancer clone(s) in multiple myeloma. Blood. 2015; 125(20): 3049–58. PubMed Abstract | Publisher Full Text | Free Full Text

15.

Walker BA, Boyle EM, Wardell CP, et al.: Mutational Spectrum, Copy Number Changes, and Outcome: Results of a Sequencing Study of Patients With Newly Diagnosed Myeloma. J Clin Oncol. 2015; 33(33): 3911–20. PubMed Abstract | Publisher Full Text | F1000 Recommendation

16.

Bolli N, Avet-Loiseau H, Wedge DC, et al.: Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat Commun. 2014; 5: 2997. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

3.

Rajkumar SV, Landgren O, Mateos MV: Smoldering multiple myeloma. Blood. 2015; 125(20): 3069–75. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

4.

Rajkumar SV, Gupta V, Fonseca R, et al.: Impact of primary molecular cytogenetic abnormalities and risk of progression in smoldering multiple myeloma. Leukemia. 2013; 27(8): 1738–44. PubMed Abstract | Publisher Full Text | Free Full Text

17.

5.

Neben K, Jauch A, Hielscher T, et al.: Progression in smoldering myeloma is independently determined by the chromosomal abnormalities del(17p), t(4;14), gain 1q, hyperdiploidy, and tumor load. J Clin Oncol. 2013; 31(34): 4325–32. PubMed Abstract | Publisher Full Text

Keats JJ, Chesi M, Egan JB, et al.: Clonal competition with alternating dominance in multiple myeloma. Blood. 2012; 120(5): 1067–76. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

18.

Kyle RA, Remstein ED, Therneau TM, et al.: Clinical course and prognosis of smoldering (asymptomatic) multiple myeloma. N Engl J Med. 2007; 356(25): 2582–90. PubMed Abstract | Publisher Full Text | F1000 Recommendation

Stewart AK, Rajkumar SV, Dimopoulos MA, et al.: Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N Engl J Med. 2015; 372(2): 142–52. PubMed Abstract | Publisher Full Text | F1000 Recommendation

19.

Hillengass J, Fechtner K, Weber MA, et al.: Prognostic significance of focal lesions in whole-body magnetic resonance imaging in patients with asymptomatic multiple myeloma. J Clin Oncol. 2010; 28(9): 1606–10. PubMed Abstract | Publisher Full Text | F1000 Recommendation

Danhof S, Schreder M, Rasche L, et al.: ‘Real-life’ experience of preapproval carfilzomib-based therapy in myeloma - analysis of cardiac toxicity and predisposing factors. Eur J Haematol. 2016; 97(1): 25–32. PubMed Abstract | Publisher Full Text | F1000 Recommendation

20.

Dispenzieri A, Kyle RA, Katzmann JA, et al.: Immunoglobulin free light chain ratio is an independent risk factor for progression of smoldering (asymptomatic) multiple myeloma. Blood. 2008; 111(2): 785–9. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

21.

González-Calle V, Dávila J, Escalante F, et al.: Bence Jones proteinuria in smoldering multiple myeloma as a predictor marker of progression to symptomatic multiple myeloma. Leukemia. 2016. PubMed Abstract | Publisher Full Text | F1000 Recommendation

Lendvai N, Hilden P, Devlin S, et al.: A phase 2 single-center study of carfilzomib 56 mg/m2 with or without low-dose dexamethasone in relapsed multiple myeloma. Blood. 2014; 124(6): 899–906. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation Kupperman E, Lee EC, Cao Y, et al.: Evaluation of the proteasome inhibitor MLN9708 in preclinical models of human cancer. Cancer Res. 2010; 70(5): 1970–80. PubMed Abstract | Publisher Full Text

22.

Mateos MV, Hernández MT, Giraldo P, et al.: Lenalidomide plus dexamethasone for high-risk smoldering multiple myeloma. N Engl J Med. 2013; 369(5): 438–47. PubMed Abstract | Publisher Full Text | F1000 Recommendation

Kumar SK, LaPlant B, Roy V, et al.: Phase 2 trial of ixazomib in patients with relapsed multiple myeloma not refractory to bortezomib. Blood Cancer J. 2015; 5: e338. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

23.

Moreau P, Masszi T, Grzasko N, et al.: Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med. 2016; 374(17): 1621–34. PubMed Abstract | Publisher Full Text | F1000 Recommendation

24.

Lu G, Middleton RE, Sun H, et al.: The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science. 2014; 343(6168): 305–9. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

25.

Krönke J, Udeshi ND, Narla A, et al.: Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science. 2014; 343(6168): 301–5. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

6.

7.

8.

9.

10.

11.

Korde N, Roschewski M, Zingone A, et al.: Treatment With CarfilzomibLenalidomide-Dexamethasone With Lenalidomide Extension in Patients With Smoldering or Newly Diagnosed Multiple Myeloma. JAMA Oncol. 2015; 1(6): 746–54. PubMed Abstract | Publisher Full Text | F1000 Recommendation

12.

Palumbo A, Avet-Loiseau H, Oliva S, et al.: Revised International Staging System for Multiple Myeloma: A Report From International Myeloma Working Group. J Clin Oncol. 2015; 33(26): 2863–9. PubMed Abstract | Publisher Full Text | Free Full Text

13.

Morgan GJ, Walker BA, Davies FE: The genetic architecture of multiple

26.

San Miguel J, Weisel K, Moreau P, et al.: Pomalidomide plus low-dose

Page 7 of 10

F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

dexamethasone versus high-dose dexamethasone alone for patients with relapsed and refractory multiple myeloma (MM-003): a randomised, open-label, phase 3 trial. Lancet Oncol. 2013; 14(11): 1055–66. PubMed Abstract | Publisher Full Text | F1000 Recommendation 27.

Leleu X, Karlin L, Macro M, et al.: Pomalidomide plus low-dose dexamethasone in multiple myeloma with deletion 17p and/or translocation (4;14): IFM 2010-02 trial results. Blood. 2015; 125(9): 1411–7. PubMed Abstract | Publisher Full Text | F1000 Recommendation

Working Group report. Blood. 2015; 125(13): 2068–74. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 45.

Kleber M, Ihorst G, Terhorst M, et al.: Comorbidity as a prognostic variable in multiple myeloma: comparative evaluation of common comorbidity scores and use of a novel MM-comorbidity score. Blood Cancer J. 2011; 1(9): e35. PubMed Abstract | Publisher Full Text | Free Full Text

46.

Garderet L, Iacobelli S, Moreau P, et al.: Superiority of the triple combination of bortezomib-thalidomide-dexamethasone over the dual combination of thalidomide-dexamethasone in patients with multiple myeloma progressing or relapsing after autologous transplantation: the MMVAR/IFM 2005-04 Randomized Phase III Trial from the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. J Clin Oncol. 2012; 30(20): 2475–82. PubMed Abstract | Publisher Full Text

28.

Morgan G, Palumbo A, Dhanasiri S, et al.: Overall survival of relapsed and refractory multiple myeloma patients after adjusting for crossover in the MM-003 trial for pomalidomide plus low-dose dexamethasone. Br J Haematol. 2015; 168(6): 820–3. PubMed Abstract | Publisher Full Text | F1000 Recommendation

29.

Fouquet G, Pegourie B, Macro M, et al.: Safe and prolonged survival with long-term exposure to pomalidomide in relapsed/refractory myeloma. Ann Oncol. 2016; 27(5): 902–7. PubMed Abstract | Publisher Full Text | F1000 Recommendation

47.

30.

Dimopoulos MA, Leleu X, Palumbo A, et al.: Expert panel consensus statement on the optimal use of pomalidomide in relapsed and refractory multiple myeloma. Leukemia. 2014; 28(8): 1573–85. PubMed Abstract | Publisher Full Text | Free Full Text

Attal M, Harousseau JL, Stoppa AM, et al.: A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Français du Myélome. N Engl J Med. 1996; 335(2): 91–7. PubMed Abstract | Publisher Full Text

48.

31.

Rosovsky R, Hong F, Tocco D, et al.: Endothelial stress products and coagulation markers in patients with multiple myeloma treated with lenalidomide plus dexamethasone: an observational study. Br J Haematol. 2013; 160(3): 351–8. PubMed Abstract | Publisher Full Text | Free Full Text

Child JA, Morgan GJ, Davies FE, et al.: High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med. 2003; 348(19): 1875–83. PubMed Abstract | Publisher Full Text

49.

Fermand JP, Ravaud P, Chevret S, et al.: High-dose therapy and autologous peripheral blood stem cell transplantation in multiple myeloma: up-front or rescue treatment? Results of a multicenter sequential randomized clinical trial. Blood. 1998; 92(9): 3131–6. PubMed Abstract

32.

Richardson PG, Schlossman RL, Alsina M, et al.: PANORAMA 2: panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myeloma. Blood. 2013; 122(14): 2331–7. PubMed Abstract | Publisher Full Text

50.

33.

San-Miguel JF, Hungria VT, Yoon SS, et al.: Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial. Lancet Oncol. 2014; 15(11): 1195–206. PubMed Abstract | Publisher Full Text | F1000 Recommendation

Mellqvist UH, Gimsing P, Hjertner O, et al.: Bortezomib consolidation after autologous stem cell transplantation in multiple myeloma: a Nordic Myeloma Study Group randomized phase 3 trial. Blood. 2013; 121(23): 4647–54. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

51.

McCarthy PL Jr, Hahn T, Hassebroek A, et al.: Trends in use of and survival after autologous hematopoietic cell transplantation in North America, 1995–2005: significant improvement in survival for lymphoma and myeloma during a period of increasing recipient age. Biol Blood Marrow Transplant. 2013; 19(17): 1116–23. PubMed Abstract | Publisher Full Text | Free Full Text

34.

Lokhorst HM, Plesner T, Laubach JP, et al.: Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma. N Engl J Med. 2015; 373(13): 1207–19. PubMed Abstract | Publisher Full Text | F1000 Recommendation

52.

35.

Lonial S, Dimopoulos M, Palumbo A, et al.: Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. N Engl J Med. 2015; 373(7): 621–31. PubMed Abstract | Publisher Full Text | F1000 Recommendation

Cavo M, Pantani L, Petrucci MT, et al.: Bortezomib-thalidomide-dexamethasone is superior to thalidomide-dexamethasone as consolidation therapy after autologous hematopoietic stem cell transplantation in patients with newly diagnosed multiple myeloma. Blood. 2012; 120(1): 9–19. PubMed Abstract | Publisher Full Text

53.

36.

Martin T, Richter J, Vij R, et al.: A Dose Finding Phase II Trial of Isatuximab (SAR650984, Anti-CD38 mAB) As a Single Agent in Relapsed/Refractory Multiple Myeloma. Blood. ASH, 2015; 126(23): 509. Reference Source

Ladetto M, Pagliano G, Ferrero S, et al.: Major tumor shrinking and persistent molecular remissions after consolidation with bortezomib, thalidomide, and dexamethasone in patients with autografted myeloma. J Clin Oncol. 2010; 28(12): 2077–84. PubMed Abstract | Publisher Full Text | F1000 Recommendation

37.

Cavo M, Tacchetti P, Patriarca F, et al.: Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet. 2010; 376(9758): 2075–85. PubMed Abstract | Publisher Full Text

54.

Paiva B, Vidriales M, Cervero J, et al.: Multiparameter flow cytometric remission is the most relevant prognostic factor for multiple myeloma patients who undergo autologous stem cell transplantation. Blood. 2008; 112(10): 4017–23. PubMed Abstract | Publisher Full Text | F1000 Recommendation

38.

Moreau P, Avet-Loiseau H, Facon T, et al.: Bortezomib plus dexamethasone versus reduced-dose bortezomib, thalidomide plus dexamethasone as induction treatment before autologous stem cell transplantation in newly diagnosed multiple myeloma. Blood. 2011; 118(22): 5752–8; quiz 5982. PubMed Abstract | Publisher Full Text

55.

Gay F, Oliva S, Petrucci MT, et al.: Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: a randomised, multicentre, phase 3 trial. Lancet Oncol. 2015; 16(16): 1617–29. PubMed Abstract | Publisher Full Text | F1000 Recommendation

39.

Durie B, Hoering A, Rajkumar SV, et al.: Bortezomib, Lenalidomide and Dexamethasone vs. Lenalidomide and Dexamethasone in Patients (Pts) with Previously Untreated Multiple Myeloma Without an Intent for Immediate Autologous Stem Cell Transplant (ASCT): Results of the Randomized Phase III Trial SWOG S0777. Blood. ASH Annual Meeting Abstracts, 2015; 126(23): 25. Reference Source

56.

Palumbo A, Cavallo F, Gay F, et al.: Autologous transplantation and maintenance therapy in multiple myeloma. N Engl J Med. 2014; 371(10): 895–905. PubMed Abstract | Publisher Full Text | F1000 Recommendation

57.

Attal M, Lauwers-Cances V, Hulin C, et al.: Autologous Transplantation for Multiple Myeloma in the Era of New Drugs: A Phase III Study of the Intergroupe Francophone Du Myelome (IFM/DFCI 2009 Trial). Blood. 2015. 126(23): 391. Reference Source

58.

McCarthy PL, Owzar K, Hofmeister CC, et al.: Lenalidomide after stem-cell transplantation for multiple myeloma. N Engl J Med. 2012; 366(19): 1770–81. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

59.

Attal M, Lauwers-Cances V, Marit G, et al.: Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. N Engl J Med. 2012; 366(19): 1782–91. PubMed Abstract | Publisher Full Text

60.

Rosiñol L, Oriol A, Teruel AI, et al.: Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study. Blood. 2012; 120(8): 1589–96. PubMed Abstract | Publisher Full Text

61.

Roussel M, Lauwers-Cances V, Robillard N, et al.: Front-line transplantation program with lenalidomide, bortezomib, and dexamethasone combination as induction and consolidation followed by lenalidomide maintenance in patients with multiple myeloma: a phase II study by the Intergroupe Francophone du Myélome. J Clin Oncol. 2014; 32(25): 2712–7. PubMed Abstract | Publisher Full Text | F1000 Recommendation

40.

Reeder CB, Reece DE, Kukreti V, et al.: Cyclophosphamide, bortezomib and dexamethasone induction for newly diagnosed multiple myeloma: high response rates in a phase II clinical trial. Leukemia. 2009; 23(7): 1337–41. PubMed Abstract | Publisher Full Text | Free Full Text

41.

Moreau P, Hulin C, Macro M, et al.: VTD is superior to VCD prior to intensive therapy in multiple myeloma: results of the prospective IFM2013-04 trial. Blood. 2016; 127(21): 2569–74. PubMed Abstract | Publisher Full Text | F1000 Recommendation

42.

Jakubowiak AJ, Dytfeld D, Griffith KA, et al.: A phase 1/2 study of carfilzomib in combination with lenalidomide and low-dose dexamethasone as a frontline treatment for multiple myeloma. Blood. 2012; 120(9): 1801–9. PubMed Abstract | Publisher Full Text | F1000 Recommendation

43.

Magarotto V, Bringhen S, Offidani M, et al.: Triplet vs doublet lenalidomidecontaining regimens for the treatment of elderly patients with newly diagnosed multiple myeloma. Blood. 2016; 127(9): 1102–8. PubMed Abstract | Publisher Full Text | F1000 Recommendation

44.

Palumbo A, Bringhen S, Mateos MV, et al.: Geriatric assessment predicts survival and toxicities in elderly myeloma patients: an International Myeloma

Page 8 of 10

F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

62.

Palumbo A, Gay F, Cavallo F, et al.: Continuous Therapy Versus Fixed Duration of Therapy in Patients With Newly Diagnosed Multiple Myeloma. J Clin Oncol. 2015; 33(30): 3459–66. PubMed Abstract | Publisher Full Text | F1000 Recommendation

63.

Tricot G, Vesole DH, Jagannath S, et al.: Graft-versus-myeloma effect: proof of principle. Blood. 1996; 87(3): 1196–8. PubMed Abstract

64.

Krishnan A, Pasquini MC, Logan B, et al.: Autologous haemopoietic stem-cell transplantation followed by allogeneic or autologous haemopoietic stem-cell transplantation in patients with multiple myeloma (BMT CTN 0102): a phase 3 biological assignment trial. Lancet Oncol. 2011; 12(13): 1195–203. PubMed Abstract | Publisher Full Text | Free Full Text

65.

Gahrton G, Iacobelli S, Bjorkstrand B, et al.: Autologous/reduced-intensity allogeneic stem cell transplantation vs autologous transplantation in multiple myeloma: long-term results of the EBMT-NMAM2000 study. Blood. 2013; 121(25): 5055–63. PubMed Abstract | Publisher Full Text

66.

67.

Giaccone L, Storer B, Patriarca F, et al.: Long-term follow-up of a comparison of nonmyeloablative allografting with autografting for newly diagnosed myeloma. Blood. 2011; 117(24): 6721–7. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation Björkstrand B, Iacobelli S, Hegenbart U, et al.: Tandem autologous/reducedintensity conditioning allogeneic stem-cell transplantation versus autologous transplantation in myeloma: long-term follow-up. J Clin Oncol. 2011; 29(22): 3016–22. PubMed Abstract | Publisher Full Text | F1000 Recommendation

73.

Rosenblatt J, Vasir B, Uhl L, et al.: Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma. Blood. 2011; 117(2): 393–402. PubMed Abstract | Publisher Full Text | Free Full Text

74.

Rapoport AP, Aqui NA, Stadtmauer EA, et al.: Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of hTERT and survivin after ASCT for myeloma. Blood. 2011; 117(3): 788–97. PubMed Abstract | Publisher Full Text | Free Full Text

75.

Lacy MQ, Mandrekar S, Dispenzieri A, et al.: Idiotype-pulsed antigen-presenting cells following autologous transplantation for multiple myeloma may be associated with prolonged survival. Am J Hematol. 2009; 84(12): 799–802. PubMed Abstract | Publisher Full Text | Free Full Text

76.

Garfall AL, Stadtmauer EA, June CH: Chimeric Antigen Receptor T Cells in Myeloma. N Engl J Med. 2016; 374(2): 194. PubMed Abstract | Publisher Full Text

77.

Benson DM Jr, Bakan CE, Mishra A, et al.: The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010; 116(13): 2286–94. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

78.

Lekoshin AM, Ansell SM, Armand P, et al.: Preliminary Results of a Phase I Study of Nivolumab (BMS-936558) in Patients with Relapsed or Refractory Lymphoid Malignancies. Blood. ASH, Abstract, 2014; 124(21): 291. Reference Source San Miguel J, Mateos MV, Shah JJ, et al.: Pembrolizumab in combination with Lenalidomide and low-dose dexamethasone for Relapsed/Refractory Multiple Myeloma (RRMM): Keynote-023. Blood. ASH, 2015; 126(23): 505. Reference Source Benson DM Jr, Hofmeister CC, Padmanabhan S, et al.: A phase 1 trial of the antiKIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma. Blood. 2012; 120(22): 4324–33. PubMed Abstract | Publisher Full Text | Free Full Text Shah N, Li L, Kaur I, et al.: Infusion of Ex Vivo Expanded Allogeneic Cord Blood-Derived Natural Killer Cells in Combination with Autologous Stem Cell Transplantation for Multiple Myeloma: Results of a Phase I Study. Blood. ASH, 2015; 126(23): 929. Reference Source

79.

68.

Bruno B, Rotta M, Patriarca F, et al.: Nonmyeloablative allografting for newly diagnosed multiple myeloma: the experience of the Gruppo Italiano Trapianti di Midollo. Blood. 2009; 113(14): 3375–82. PubMed Abstract | Publisher Full Text | Free Full Text

80.

69.

Lokhorst HM, Schattenberg A, Cornelissen JJ, et al.: Donor lymphocyte infusions for relapsed multiple myeloma after allogeneic stem-cell transplantation: predictive factors for response and long-term outcome. J Clin Oncol. 2000; 18(16): 3031–7. PubMed Abstract

81.

70.

Noonan KA, Huff CA, Davis J, et al.: Adoptive transfer of activated marrowinfiltrating lymphocytes induces measurable antitumor immunity in the bone marrow in multiple myeloma. Sci Transl Med. 2015; 7(288): 288ra78. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

82.

71.

Rosenblatt J, Glotzbecker B, Mills H, et al.: PD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses to autologous dendritic cell/ myeloma fusion vaccine. J Immunother. 2011; 34(5): 409–18. PubMed Abstract | Publisher Full Text | Free Full Text

83.

72.

Garfall AL, Fraietta JA, Maus MV: Immunotherapy with chimeric antigen receptors for multiple myeloma. Discov Med. 2014; 17(91): 37–46. PubMed Abstract

84.

Szmania S, Lapteva N, Garg T, et al.: Ex vivo-expanded natural killer cells demonstrate robust proliferation in vivo in high-risk relapsed multiple myeloma patients. J Immunother. 2015; 38(1): 24–36. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation Porrata LF, Gertz MA, Inwards DJ, et al.: Early lymphocyte recovery predicts superior survival after autologous hematopoietic stem cell transplantation in multiple myeloma or non-Hodgkin lymphoma. Blood. 2001; 98(3): 579–85. PubMed Abstract | Publisher Full Text Barlogie B, Mitchell A, van Rhee F, et al.: Curing myeloma at last: defining criteria and providing the evidence. Blood. 2014; 124(20): 3043–51. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation

Page 9 of 10

F1000Research 2016, 5(F1000 Faculty Rev):2053 Last updated: 23 AUG 2016

Open Peer Review Current Referee Status: Editorial Note on the Review Process F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).

The referees who approved this article are: Version 1 1 David Avigan, Beth Israel Deaconess Medical Center & Harvard Medical School, Boston, Massachusetts, USA Competing Interests: No competing interests were disclosed. 2 Qaiser Bashir, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA Competing Interests: No competing interests were disclosed. 3 Stefan Knop, Wilhelm Sander Myeloma Unit, Division of Hematology and Medical Oncology, Wuerzburg University Medical Center, Wuerzburg, Germany Competing Interests: No competing interests were disclosed. 4 Monika Engelhardt, Department of Hematology and Oncology, University of Freiburg Medical Center, Freiburg, Germany Competing Interests: No competing interests were disclosed.

F1000Research Page 10 of 10

Recent advances in understanding multiple myeloma.

There have been major recent advancements in the understanding and management of multiple myeloma. Diagnostic criteria have been revised and former ul...
1001KB Sizes 1 Downloads 13 Views